Review

The role of PSMD9 in human disease: future clinical and therapeutic implications

  • Received: 22 October 2015 Accepted: 25 November 2015 Published: 25 January 2015
  • PSMD9 was first characterized as a component of the PA700 proteasomal regulator, and was found to stimulate association of PA700 with the catalytic 20S proteasomal core to form the active 26S proteasome. It was also independently identified under the name “bridge-1” as a transcriptional co-activator that modulates function of the transcription factors PDX-1, E12, and E47, and interacts with the co-activator histone acetyltransferase p300. Here, we discuss the molecular and genetic data linking PSMD9 to a diverse range of conditions including diabetes, cancer, mental health problems, polycystic ovary syndrome and neurodegenerative diseases, and thereby highlight its potential as a therapeutic target in these multiple settings.

    Citation: Joanne L. Hopper, Natasha Begum, Laura Smith, Thomas A. Hughes. The role of PSMD9 in human disease: future clinical and therapeutic implications[J]. AIMS Molecular Science, 2015, 2(4): 476-484. doi: 10.3934/molsci.2015.4.476

    Related Papers:

    [1] Kazunobu Baba, Tadaaki Miyazaki . Critical function of Siah2 in tumorigenesis. AIMS Molecular Science, 2017, 4(4): 415-423. doi: 10.3934/molsci.2017.4.415
    [2] Kenji Chiba, Yasuhiro Maeda, Noriyasu Seki, Hirotoshi Kataoka, Kunio Sugahara . Role of sphingosine 1-phosphate (S1P) and effects of fingolimod, an S1P receptor 1 functional antagonist in lymphocyte circulation and autoimmune diseases. AIMS Molecular Science, 2014, 1(4): 162-182. doi: 10.3934/molsci.2014.4.162
    [3] Andrew V. Nguyen, Yuan-Yuan Wu, Elaine Y. Lin . The regulatory function of sphingosine-1-phosphate signaling axis on regulatory T cells in colorectal cancer. AIMS Molecular Science, 2015, 1(1): 34-47. doi: 10.3934/molsci.2015.1.34
    [4] Leena Latonen . Protein aggregation in neurodegenerative disease: the nucleolar connection. AIMS Molecular Science, 2015, 2(3): 324-331. doi: 10.3934/molsci.2015.3.324
    [5] Sayeeda Ahsanuddin, Minh Lam, Elma D. Baron . Skin aging and oxidative stress. AIMS Molecular Science, 2016, 3(2): 187-195. doi: 10.3934/molsci.2016.2.187
    [6] Seidu A. Richard, Wu Min, Zhaoliang Su, Hua-Xi Xu . Epochal neuroinflammatory role of high mobility group box 1 in central nervous system diseases. AIMS Molecular Science, 2017, 4(2): 185-218. doi: 10.3934/molsci.2017.2.185
    [7] Vera Reitsema, Hjalmar Bouma, Jan Willem Kok . Sphingosine-1-phosphate transport and its role in immunology. AIMS Molecular Science, 2014, 1(4): 183-201. doi: 10.3934/molsci.2014.4.183
    [8] Simone Leggeri, Navid Sobhani . Single nucleotide polymorphisms Rs1045642 C>T genetic alteration in ATP Binding Cassette Subfamily B Member 1 role in increasing everolimus toxicity in metastatic breast cancer. AIMS Molecular Science, 2020, 7(1): 1-11. doi: 10.3934/molsci.2020001
    [9] Mohamed A. Eldeeb . Aging: when the ubiquitin–proteasome machinery collapses. AIMS Molecular Science, 2017, 4(2): 219-223. doi: 10.3934/molsci.2017.2.219
    [10] Shizuka Takaku, Naoko Niimi, Toshihiko Kadoya, Hideji Yako, Masami Tsukamoto, Kunihiko Sakumi, Yusaku Nakabeppu, Hidenori Horie, Kazunori Sango . Galectin-1 and galectin-3 as key molecules for peripheral nerve degeneration and regeneration. AIMS Molecular Science, 2016, 3(3): 325-337. doi: 10.3934/molsci.2016.3.325
  • PSMD9 was first characterized as a component of the PA700 proteasomal regulator, and was found to stimulate association of PA700 with the catalytic 20S proteasomal core to form the active 26S proteasome. It was also independently identified under the name “bridge-1” as a transcriptional co-activator that modulates function of the transcription factors PDX-1, E12, and E47, and interacts with the co-activator histone acetyltransferase p300. Here, we discuss the molecular and genetic data linking PSMD9 to a diverse range of conditions including diabetes, cancer, mental health problems, polycystic ovary syndrome and neurodegenerative diseases, and thereby highlight its potential as a therapeutic target in these multiple settings.


    1. Introduction

    PSMD9 (26S Proteasome Non-ATPase Regulatory Subunit 9; also known as bridge-1, p27 or Rpn4) is a 25kDa protein encoded by the PSMD9 gene on chromosome 12q24.31-q24.32. Also known in humans as p27 (not to be confused with p27KIP1), the protein is highly conserved in model organisms such as rodents (in which it is often referred to as bridge-1), yeast (nas2) and nematodes (PSMD9). PSMD9 is ubiquitously expressed in mammalian cell types and appears to be at particularly high levels in lymphatic, endocrine, kidney, and skin epidermis tissues [1].

    PSMD9 was first characterised though its interaction with the proteasomal chaperones PSMC6 and PSMC3, with which it forms a complex that modulates the assembly of the 26S proteasome from the constituent 20S catalytic and 19S (PA700) regulatory subunits (Figure 1) [2]. In addition to this proteasomal role, it is also apparent that PSMD9 has a separate role as a transcriptional co-regulator although this role was first described under the alternative name of bridge-1. PSMD9 regulates gene expression of specific target genes by binding of its PDZ domain to the transcription factors PDX-1, E12 and E47 [3,4]. A comprehensive 3D structure of PSMD9 is yet to be elucidated but analyses of the secondary structure have revealed the presence of a unique PDZ domain, a motif associated with specific protein-protein interactions, and it is assumed that this motif underpins both functional roles. The regulatory impact of PSMD9 on both proteasomal activity and transcription remains unclear. The ubiquitin/proteasome pathway is the main non-lysosomal route for intracellular protein degradation and is responsible for the turnover of > 80% of cellular proteins [5]. Despite its role in proteasome assembly, it is unknown whether changes in PSMD9 expression impact on overall proteasomal activity, although it has been reported that PSMD9 mediates the specific proteasomal degradation of IκBα leading to regulation of NF-κB signalling [6]. Similarly, the range of transcriptional targets for the transcriptional co-activator role of PSMD9 is unknown, with experimentally confirmed targets limited to the insulin gene at this time [4]. PSMD9 has, however, been linked with a wide range of human diseases and may, in some contexts, present a novel therapeutic target.

    Figure 1. The 26S proteasome. PSMD9 is a constituent of the 19S regulatory, non-ATPase, particle that binds to the 20S core particle to assemble the complete 26S complex.

    2. PSMD9 in cancer

    Banz et al. were the first to comment on the potential relevance of PSMD9 in cancer [7]. They demonstrated that treatment of breast cancer cell lines with the TGFβ family ligand Activin A stimulated PSMD9 expression together with increased levels of the signal transduction proteins Smad2, Smad3, and Smad4. PSMD9 knock-down was also associated with decreases in the levels of these Smads, implicating PSMD9 in the transmission of Activin A signalling through the regulation of Smad proteins. Activin A has roles in various potentially cancer-related pathways including those controlling inflammation, immunity, wound repair [8] and glucose metabolism [9], and has been implicated in pro- and anti-carcinogenic roles in several tissue types [10,11,12]. The team concluded that PSMD9 is a potential regulator of cell cycle progression. Further to this, we have recently published that low expression of PSMD9 within breast cancer tissues significantly associates with reduced incidence of local recurrence in patients receiving adjuvant radiotherapy but not in those treated without [13]. There is currently no standardized methodology by which to assess the radio-sensitivity of a tumour before treatment, although potential molecular markers have been reported, including p53 [14], Bcl-2 [15] and survivin [16]. Notably, we used a clinically routine immunohistochemistry methodology for our assessment, therefore identifying PSMD9 as a clinically-translatable biomarker of radiotherapy response. We also showed that in vitro depletion of PSMD9 in breast cancer cell lines acts to sensitize cells to radiotherapy, indicating that PSMD9 may have a direct role in cellular response to radiotherapy and may represent a novel target for radio-sensitizing drugs. As yet, it is unclear whether this influence of PSMD9 relates to its role in the proteasome or as a transcriptional co-regulator, however data showing the proteasome inhibitor Bortezomib to induce similar radio-sensitization [17] imply it may be the former.

    3. PSMD9 in type II diabetes

    3.1. Genetics

    Genome-wide linkage studies have identified a type 2 diabetes (T2D) locus called “non-insulin-dependent diabetes mellitus” (NIDDM) 2 [18,19]. The PSMD9 gene lies within the NIDDM2 locus and SNPs within the PSMD9 gene have been linked to the onset of T2D [20,21]. In a fine-mapping study Gragnoli and Cronsell performed direct sequencing in 237 Italian T2D patients and uncovered four rare PSMD9 mutations/variants that were associated with the disease [21]. Significant linkage of the PSMD9 SNPs rs74421874, rs3825172 and rs1043307/rs2514259 was later identified in late onset T2D [20] as well as early-onset T2D/maturity-onset diabetes of the young (MODY) type 3 [22] in Italian families. These PSMD9 SNPs have also been significantly linked to particular pathological features of this disease including T2D-neuropathy [23], microvascular [24] and macrovascular pathology [25] and hypertension [26]. The impact of these SNPs on PSMD9 function or expression has not yet been determined.

    3.2. Pathogenesis

    Thomas et al demonstrated that PSMD9 interacted with the basic helix-loop-helix (BHLH) transcription factors E12 and E47 via its PDZ domain and increased activation of enhancers of glucose-response within the insulin promoter. Functional investigations showed that increased expression of PSMD9 promoted pancreatic β-cell survival in vivo [4,27,28] whereas decreased expression of endogenous PSMD9 in rat insulinoma (INS-1) cells diminished insulin promoter activity suggesting that reduced expression of this protein may be linked with diabetes development [4]. In contrast, Volinic et al. found that increased expression of pancreatic PSMD9 led to insulin deficiency and diabetes in a murine model, and proposed a biphasic pattern of insulin promoter activation to explain their findings [29]. They showed that increasing levels of exogenous PSMD9 resulted in a shift from transcriptional activation to repression of the insulin promoter, and suggested that this may be the result of an incremental dosage effect in which increased levels first promote but then uncouple the optimal stoichiometric assembly of transcriptional regulatory complexes [29]. This suggested that individuals with reduced or augmented PSMD9 expression levels could be at an increased risk of diabetes and that modulation of this molecule may provide an effective therapeutic strategy.

    It is now established that the inflammatory response is a major contributing pathway in both the development and progression of T2D. Chronic inflammation is triggered by the metabolic stress associated with hyperglycaemia in pancreatic islets that may lead to defective insulin secretion or response [30]. A study by Liu et al. into the role of the proteasome in diabetes demonstrated that the production of oxidative species due to hyperglycaemic conditions in diabetic models led to an increase in proteasome function in a PA700 dependant manner and that this caused subsequent activation of NF-κB [31], providing a further pointer for a mechanism of PSMD9’s influence as both a component of the PA700 complex and a known regulator of NF-κB.

    4. PSMD9 in mental health disorders

    4.1. Genetics

    Major depressive disorder (MDD) and generalised anxiety disorder (GAD) are the most prevalent mental health disorders in the UK, with approximately 1 in 5 people affected by these disorders in their lifetime. Familial connections have been indicated in these conditions, with heritability values of 30-40% [32]. Similarly, the rarer condition of Schizophrenia (SCZ), which affects approximately 1 in 100 individuals during their lifetimes [33], has a remarkably high heritability value of up to 80% [34], suggesting strong genetic contributions to the pathogenesis of these mental health illnesses. In addition, frequent connections have been made between T2D and mental health issues, with each an established comorbidity of the other [35,36]. Due to this, Gragnoli et al. further investigated the PSMD9 SNPs from their previous T2D studies and found that rs74421874, rs3825172 and rs1043307/rs2514259 were also significantly associated with both GAD [37] and MDD [38] development in T2D backgrounds. In support of this, a number of other studies have also linked PSMD9 SNPs to mental health disorders [39,40,41]. A number of PSMD9 SNPs has also been demonstrated to associate with individuals with clinically diagnosed cases of SCZ [41] and MDD [40]. Furthermore, PSMD9 rs1043307 has been correlated with a more positive response to tricyclic antidepressants [39]. This study predicted a link between PSMD9 and BHLH transcription factor binding pathway, again suggesting a possible transcriptional co-activator role for the protein, this time in the context of mental health. These data suggest that SNPs in PSMD9 may be used to identify individuals with increased risk of developing MDD or GAD in T2D patients or SCZ in the general population.

    4.2. Pathogenesis

    A definitive understanding of the molecular pathogenesis of these mental health disorders remains frustratingly evasive. Current theory focuses on dysregulation of various important proteins within the brain including stress hormones, cytokines and monoamines which may lead to decreased neuronal activity or inappropriate activation of inflammatory pathways [42,43,44,45]. In particular, a decrease in the expression of several specific proteins has been associated with SCZ with the most prominent example being the protein dysbindin [46,47,48,49]. Han et al. recently demonstrated that PSMD9 and dysbindin could be co-immunoprecipitated from mouse neuronal tissue [50], a direct interaction that supports a role for PSMD9 analogous to that previously discussed with respect to the degradation of Iκβα, in which PSMD9 may act to enhance delivery of dysbindin to the proteasome for degradation. Similarly, as part of their study into antidepressant treatment response, Wong et al. proposed that the PSMD9 SNP rs1043307 may be associated with a change in antigen processing or degradation that may lead to an alteration in the immune response of those individuals who responded more positively to anti-depressant treatments in comparison to those who do not [39]. Gragnoli et al. added to this with the suggestion that PSMD9, as a component of the 26S proteasome, may be involved in the immune-tolerance checking process [38] and dysregulation of this pathway through changes in proteasome-associated PSMD9 may lead to an autoimmune response that could trigger these mental health conditions.

    5. PSMD9 in other diseases

    5.1. Polycystic ovary syndrome

    Polycystic ovary syndrome (PCOS) is the most common endocrinopathy in women of reproductive age. Polycystic ovaries are generally enlarged and contain multiple follicles that do not undergo cyclic luteal differentiation, increasing infertility rates. Previous to their described work in breast cancer, Banz et el. examined PSMD9 function in ovary cells [51]. They performed in vitro experiments treating granulosa cells with activin A, and showed the same trends as those in cancer cells, with an increase in PSMD9 levels following activin A treatment and a decrease in Smad levels following PSMD9 knock down. The team suggested that PSMD9 may perform a regulatory role in granulosa cell proliferation and differentiation. Interestingly, PCOS demonstrates frequent correlations with T2D [52] and in many women raised levels of testosterone are indicative of the condition, which is reportedly triggered by hyperinsulinemia following a reduction in insulin sensitivity [53]. Therefore, through its relationship with the insulin signalling and activin A pathways, it is plausible that PSMD9 may become a potentially interesting new target in the treatment of PCOS and associated infertility, once data in humans with these disorders have been established.

    5.2. Neurodegeneration

    PSMD9 has been associated with a group of chronic neurodegenerative diseases broadly termed synucleinopathies, which include Parkinson’s disease and multiple system atrophy [54]. These diseases are characterised by the presence of amyloid fibrils containing α-synuclein (α-Syn) in neuronal cells [55,56,57]. Over-expression of α-Syn in the dopaminergic neurons of the model organism C. elegans resulted in an up-regulation of PSMD9 expression [58]. The authors conclude that proteasome-associated PSMD9 is likely to be a component of the α-Syn degradation pathway that may have an important role in all synucleinopathies. Indeed, proteasomal disruptions have been strongly implicated in the formation of α-Syn aggregates [59,60] and although the mechanisms remain unclear, depletion of the 26S proteasome in transgenic mice has led to the development of Parkinson’s disease-like symptoms [61]. Further to this, aggregation of α-Syn is attributed to misfolding of the protein and Sangith et al. speculated that PSMD9 may have the capacity to perform quality control of protein misfolding. They suggest that proteasome associated PSMD9 located on the endoplasmic reticulum would have the ability to bind directly to the C-terminal motifs of exiting proteins via its PDZ domain, offering a way to circumvent the ubiquitination pathway [62]. Interestingly, although α-Syn has traditionally been studied within the brain, this protein is also present in pancreatic beta cells where it is proposed to have a regulatory role in insulin secretion [63,64] and might well be involved in the development of T2D. Epidemiological data suggests that T2D may be a risk factor in the development of PD [65] and may be associated with a more severe form of the disease [66] and, although conclusive data are limited, it has been suggested that 50-80% of PD patients may have abnormal glucose tolerance [67,68]. Through its association with metabolism and proteostasis PSMD9 may therefore provide a novel avenue of research for ageing related neurodegenerative diseases.

    6. Conclusions

    PSMD9 is a component of the 26S proteasome; a mediator of protein turnover, and has been shown to act as a transcriptional co-activator. PSMD9 has been associated with the pathogenic pathways of several important human disorders. Commonalities in the pathways regulating these disorders include changes in insulin secretion, alterations in protein turnover and immune dysregulation. Here, we suggest that targeting PSMD9 might be a novel therapy for the treatment of a range of diseases.

    Conflicts of interest

    All authors declare no conflicts of interest in this paper.

    [1] Uhlen M, Oksvold P, Fagerberg L, et al. (2010) Towards a knowledge-based Human Protein Atlas. Nat Biotechnol 28: 1248-1250. doi: 10.1038/nbt1210-1248
    [2] Watanabe TK, Saito A, Suzuki M, et al. (1998) cDNA cloning and characterization of a human proteasomal modulator subunit, p27 (PSMD9). Genomics 50: 241-250. doi: 10.1006/geno.1998.5301
    [3] Stanojevic V, Yao KM, Thomas MK (2005) The coactivator Bridge-1 increases transcriptional activation by pancreas duodenum homeobox-1 (PDX-1). Mol Cell Endocrinol 237: 67-74. doi: 10.1016/j.mce.2005.03.003
    [4] Thomas MK, Yao KM, Tenser MS, et al. (1999) Bridge-1, a novel PDZ-domain coactivator of E2A-mediated regulation of insulin gene transcription. Mol Cell Biol 19: 8492-8504. doi: 10.1128/MCB.19.12.8492
    [5] Dalton WS (2004) The proteasome. Semin Oncol 31: 3-9. doi: 10.1053/S0093-7754(04)00498-1
    [6] Sahu I, Sangith N, Ramteke M, et al. (2014) A novel role for the proteasomal chaperone PSMD9 and hnRNPA1 in enhancing IkappaBalpha degradation and NF-kappaB activation - functional relevance of predicted PDZ domain-motif interaction. FEBS J 281: 2688-2709. doi: 10.1111/febs.12814
    [7] Banz-Jansen C, Munchow B, Diedrich K, et al. (2011) Bridge-1 is expressed in human breast carcinomas: silencing of Bridge-1 decreases Smad2, Smad3 and Smad4 expression in MCF-7 cells, a human breast cancer cell line. Arch Gynecol Obstet 284: 1543-1549. doi: 10.1007/s00404-011-1875-0
    [8] de Kretser DM, O'Hehir RE, Hardy CL, et al. (2012) The roles of activin A and its binding protein, follistatin, in inflammation and tissue repair. Mol Cell Endocrinol 359: 101-106. doi: 10.1016/j.mce.2011.10.009
    [9] Hashimoto O, Funaba M (2011) Activin in glucose metabolism. Vitam Horm 85: 217-234. doi: 10.1016/B978-0-12-385961-7.00011-1
    [10] Deli A, Kreidl E, Santifaller S, et al. (2008) Activins and activin antagonists in hepatocellular carcinoma. World J Gastroenterol 14: 1699-1709. doi: 10.3748/wjg.14.1699
    [11] Ottley E, Gold E (2012) Insensitivity to the growth inhibitory effects of activin A: an acquired capability in prostate cancer progression. Cytokine Growth Factor Rev 23: 119-125. doi: 10.1016/j.cytogfr.2012.04.004
    [12] Loomans HA, Andl CD (2014) Intertwining of Activin A and TGFbeta Signaling: Dual Roles in Cancer Progression and Cancer Cell Invasion. Cancers (Basel) 7: 70-91. doi: 10.3390/cancers7010070
    [13] Langlands FE, Dodwell D, Hanby AM, et al. (2014) PSMD9 expression predicts radiotherapy response in breast cancer. Mol Cancer 13: 73. doi: 10.1186/1476-4598-13-73
    [14] Alsner J, Sorensen SB, Overgaard J (2001) TP53 mutation is related to poor prognosis after radiotherapy, but not surgery, in squamous cell carcinoma of the head and neck. Radiother Oncol 59: 179-185. doi: 10.1016/S0167-8140(01)00301-2
    [15] Abdel Raheem AM, Hameed DA, ElGanainy EO, et al. (2011) Can Bcl-XL expression predict the radio sensitivity of bilharzial-related squamous bladder carcinoma? A prospective comparative study. BMC Cancer 11: 16.
    [16] Asanuma K, Moriai R, Yajima T, et al. (2000) Survivin as a radioresistance factor in pancreatic cancer. Jpn J Cancer Res 91: 1204-1209. doi: 10.1111/j.1349-7006.2000.tb00906.x
    [17] Crawford LJ, Walker B, Irvine AE (2011) Proteasome inhibitors in cancer therapy. J Cell Commun Signal 5: 101-110. doi: 10.1007/s12079-011-0121-7
    [18] Hanis CL, Boerwinkle E, Chakraborty R, et al. (1996) A genome-wide search for human non-insulin-dependent (type 2) diabetes genes reveals a major susceptibility locus on chromosome 2. Nat Genet 13: 161-166. doi: 10.1038/ng0696-161
    [19] Mahtani MM, Widen E, Lehto M, et al. (1996) Mapping of a gene for type 2 diabetes associated with an insulin secretion defect by a genome scan in Finnish families. Nat Genet 14: 90-94. doi: 10.1038/ng0996-90
    [20] Gragnoli C (2010) PSMD9 gene in the NIDDM2 locus is linked to type 2 diabetes in Italians. J Cell Physiol 222: 265-267. doi: 10.1002/jcp.21954
    [21] Gragnoli C, Cronsell J (2007) PSMD9 gene variants within NIDDM2 may rarely contribute to type 2 diabetes. J Cell Physiol 212: 568-571. doi: 10.1002/jcp.21127
    [22] Gragnoli C (2010) PSMD9 is linked to MODY3. J Cell Physiol 223: 1-5.
    [23] Gragnoli C (2011) PSMD9 is linked to type 2 diabetes neuropathy. J Diabetes Complications 25: 329-331. doi: 10.1016/j.jdiacomp.2011.06.003
    [24] Gragnoli C (2012) Proteasome modulator 9 is linked to microvascular pathology of T2D. J Cell Physiol 227: 3116-3118. doi: 10.1002/jcp.23063
    [25] Gragnoli C (2011) Proteasome modulator 9 and macrovascular pathology of T2D. Cardiovasc Diabetol 10: 32. doi: 10.1186/1475-2840-10-32
    [26] Gragnoli C (2011) Proteasome modulator 9 SNPs are linked to hypertension in type 2 diabetes families. Cardiovasc Diabetol 10: 77. doi: 10.1186/1475-2840-10-77
    [27] Lee JH, Volinic JL, Banz C, et al. (2005) Interactions with p300 enhance transcriptional activation by the PDZ-domain coactivator Bridge-1. J Endocrinol 187: 283-292. doi: 10.1677/joe.1.06305
    [28] Thomas MK, Tsang SW, Yeung ML, et al. (2009) The roles of the PDZ-containing proteins bridge-1 and PDZD2 in the regulation of insulin production and pancreatic beta-cell mass. Curr Protein Pept Sci 10: 30-36. doi: 10.2174/138920309787315248
    [29] Volinic JL, Lee JH, Eto K, et al. (2006) Overexpression of the coactivator bridge-1 results in insulin deficiency and diabetes. Mol Endocrinol 20: 167-182. doi: 10.1210/me.2005-0127
    [30] Donath MY (2014) Targeting inflammation in the treatment of type 2 diabetes: time to start. Nat Rev Drug Discov 13: 465-476. doi: 10.1038/nrd4275
    [31] Liu H, Yu S, Xu W, et al. (2012) Enhancement of 26S proteasome functionality connects oxidative stress and vascular endothelial inflammatory response in diabetes mellitus. Arterioscler Thromb Vasc Biol 32: 2131-2140. doi: 10.1161/ATVBAHA.112.253385
    [32] Sullivan PF, Neale MC, Kendler KS (2000) Genetic epidemiology of major depression: review and meta-analysis. Am J Psychiatry 157: 1552-1562. doi: 10.1176/appi.ajp.157.10.1552
    [33] Millier A, Schmidt U, Angermeyer MC, et al. (2014) Humanistic burden in schizophrenia: a literature review. J Psychiatr Res 54: 85-93. doi: 10.1016/j.jpsychires.2014.03.021
    [34] Cardno AG, Marshall EJ, Coid B, et al. (1999) Heritability estimates for psychotic disorders: the Maudsley twin psychosis series. Arch Gen Psychiatry 56: 162-168. doi: 10.1001/archpsyc.56.2.162
    [35] Mezuk B, Eaton WW, Albrecht S, et al. (2008) Depression and type 2 diabetes over the lifespan: a meta-analysis. Diabetes Care 31: 2383-2390. doi: 10.2337/dc08-0985
    [36] Pan A, Lucas M, Sun Q, et al. (2010) Bidirectional association between depression and type 2 diabetes mellitus in women. Arch Intern Med 170: 1884-1891.
    [37] Gragnoli C (2014) Proteasome modulator 9 gene SNPs, responsible for anti-depressant response, are in linkage with generalized anxiety disorder. J Cell Physiol 229: 1157-1159. doi: 10.1002/jcp.24581
    [38] Gragnoli C (2012) Proteasome modulator 9 and depression in type 2 diabetes. Curr Med Chem 19: 5178-5180. doi: 10.2174/092986712803530593
    [39] Wong ML, Dong C, Maestre-Mesa J, et al. (2008) Polymorphisms in inflammation-related genes are associated with susceptibility to major depression and antidepressant response. Mol Psychiatry 13: 800-812. doi: 10.1038/mp.2008.59
    [40] Wong ML, Dong C, Andreev V, et al. (2012) Prediction of susceptibility to major depression by a model of interactions of multiple functional genetic variants and environmental factors. Mol Psychiatry 17: 624-633. doi: 10.1038/mp.2012.13
    [41] Lee YH, Kim JH, Song GG (2013) Pathway analysis of a genome-wide association study in schizophrenia. Gene 525: 107-115. doi: 10.1016/j.gene.2013.04.014
    [42] Haase J, Brown E (2015) Integrating the monoamine, neurotrophin and cytokine hypotheses of depression--a central role for the serotonin transporter? Pharmacol Ther 147: 1-11. doi: 10.1016/j.pharmthera.2014.10.002
    [43] Furtado M, Katzman MA (2015) Examining the role of neuroinflammation in major depression. Psychiatry Res 229: 27-36. doi: 10.1016/j.psychres.2015.06.009
    [44] Uzbekov M, Maxinova N (2015) Biochemical Bases of Monoamine and Hormonal Interactions in Pathogenesis of Anxious Depression: a Hypothesis. European Psychiatry 30: 542.
    [45] Najjar S, Pearlman DM (2015) Neuroinflammation and white matter pathology in schizophrenia: systematic review. Schizophr Res 161: 102-112. doi: 10.1016/j.schres.2014.04.041
    [46] Talbot K, Eidem WL, Tinsley CL, et al. (2004) Dysbindin-1 is reduced in intrinsic, glutamatergic terminals of the hippocampal formation in schizophrenia. J Clin Invest 113: 1353-1363. doi: 10.1172/JCI200420425
    [47] Weickert CS, Rothmond DA, Hyde TM, et al. (2008) Reduced DTNBP1 (dysbindin-1) mRNA in the hippocampal formation of schizophrenia patients. Schizophr Res 98: 105-110.
    [48] Saggu S, Cannon TD, Jentsch JD, et al. (2013) Potential molecular mechanisms for decreased synaptic glutamate release in dysbindin-1 mutant mice. Schizophr Res 146: 254-263. doi: 10.1016/j.schres.2013.01.037
    [49] Tang J, LeGros RP, Louneva N, et al. (2009) Dysbindin-1 in dorsolateral prefrontal cortex of schizophrenia cases is reduced in an isoform-specific manner unrelated to dysbindin-1 mRNA expression. Hum Mol Genet 18: 3851-3863. doi: 10.1093/hmg/ddp329
    [50] Han MH, Hu Z, Chen CY, et al. (2014) Dysbindin-associated proteome in the p2 synaptosome fraction of mouse brain. J Proteome Res 13: 4567-4580. doi: 10.1021/pr500656z
    [51] Banz C, Munchow B, Diedrich K (2010) Bridge-1 is expressed in human granulosa cells and is involved in the activin A signaling cascade. Fertil Steril 93: 1349-1352. doi: 10.1016/j.fertnstert.2009.07.1675
    [52] Barber TM, Franks S (2012) The link between polycystic ovary syndrome and both Type 1 and Type 2 diabetes mellitus: what do we know today? Womens Health (Lond Engl) 8: 147-154. doi: 10.2217/whe.11.94
    [53] Ehrmann DA (2005) Polycystic ovary syndrome. N Engl J Med 352: 1223-1236. doi: 10.1056/NEJMra041536
    [54] Marti MJ, Tolosa E, Campdelacreu J (2003) Clinical overview of the synucleinopathies. Mov Disord 18 Suppl 6: S21-27.
    [55] Stefanova N, Klimaschewski L, Poewe W, et al. (2001) Glial cell death induced by overexpression of alpha-synuclein. J Neurosci Res 65: 432-438. doi: 10.1002/jnr.1171
    [56] Xilouri M, Brekk OR, Stefanis L (2013) alpha-Synuclein and protein degradation systems: a reciprocal relationship. Mol Neurobiol 47: 537-551. doi: 10.1007/s12035-012-8341-2
    [57] Stefanis L (2012) alpha-Synuclein in Parkinson's disease. Cold Spring Harb Perspect Med 2: a009399.
    [58] Vartiainen S, Pehkonen P, Lakso M, et al. (2006) Identification of gene expression changes in transgenic C. elegans overexpressing human alpha-synuclein. Neurobiol Dis 22: 477-486.
    [59] Rideout HJ, Dietrich P, Wang Q, et al. (2004) alpha-synuclein is required for the fibrillar nature of ubiquitinated inclusions induced by proteasomal inhibition in primary neurons. J Biol Chem 279: 46915-46920. doi: 10.1074/jbc.M405146200
    [60] Pierre S-R, Vernace V, Wang Z, et al. (2009) Mechanisms Linking the Ubiquitin/Proteasome Pathway and Chaperones. In: Richter-Landsberg C, editor. Heat Shock Proteins in Neural Cells: Springer New York.
    [61] Bedford L, Hay D, Devoy A, et al. (2008) Depletion of 26S proteasomes in mouse brain neurons causes neurodegeneration and Lewy-like inclusions resembling human pale bodies. J Neurosci 28: 8189-8198. doi: 10.1523/JNEUROSCI.2218-08.2008
    [62] Sangith N, Srinivasaraghavan K, Sahu I, et al. (2014) Discovery of novel interacting partners of PSMD9, a proteasomal chaperone: Role of an Atypical and versatile PDZ-domain motif interaction and identification of putative functional modules. FEBS Open Bio 4: 571-583. doi: 10.1016/j.fob.2014.05.005
    [63] Geng X, Lou H, Wang J, et al. (2011) alpha-Synuclein binds the K(ATP) channel at insulin-secretory granules and inhibits insulin secretion. Am J Physiol Endocrinol Metab 300: E276-286. doi: 10.1152/ajpendo.00262.2010
    [64] Steneberg P, Bernardo L, Edfalk S, et al. (2013) The type 2 diabetes-associated gene ide is required for insulin secretion and suppression of alpha-synuclein levels in beta-cells. Diabetes 62: 2004-2014. doi: 10.2337/db12-1045
    [65] Hu G, Jousilahti P, Bidel S, et al. (2007) Type 2 diabetes and the risk of Parkinson's disease. Diabetes Care 30: 842-847. doi: 10.2337/dc06-2011
    [66] Cereda E, Barichella M, Cassani E, et al. (2012) Clinical features of Parkinson disease when onset of diabetes came first: A case-control study. Neurology 78: 1507-1511. doi: 10.1212/WNL.0b013e3182553cc9
    [67] Cereda E, Barichella M, Pedrolli C, et al. (2011) Diabetes and risk of Parkinson's disease: a systematic review and meta-analysis. Diabetes Care 34: 2614-2623. doi: 10.2337/dc11-1584
    [68] Sandyk R (1993) The relationship between diabetes mellitus and Parkinson's disease. Int J Neurosci 69: 125-130. doi: 10.3109/00207459309003322
  • This article has been cited by:

    1. Adrien Rousseau, Anne Bertolotti, Regulation of proteasome assembly and activity in health and disease, 2018, 19, 1471-0072, 697, 10.1038/s41580-018-0040-z
    2. Peter Larsson, Daniella Pettersson, Hanna Engqvist, Elisabeth Werner Rönnerman, Eva Forssell-Aronsson, Anikó Kovács, Per Karlsson, Khalil Helou, Toshima Z. Parris, Pan-cancer analysis of genomic and transcriptomic data reveals the prognostic relevance of human proteasome genes in different cancer types, 2022, 22, 1471-2407, 10.1186/s12885-022-10079-4
  • Reader Comments
  • © 2015 the Author(s), licensee AIMS Press. This is an open access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/4.0)
通讯作者: 陈斌, bchen63@163.com
  • 1. 

    沈阳化工大学材料科学与工程学院 沈阳 110142

  1. 本站搜索
  2. 百度学术搜索
  3. 万方数据库搜索
  4. CNKI搜索

Metrics

Article views(6744) PDF downloads(1408) Cited by(2)

Article outline

/

DownLoad:  Full-Size Img  PowerPoint
Return
Return

Catalog